<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Digital.CSIC

MINI REVIEW published: 30 January 2019 doi: 10.3389/fmed.2019.00010

Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function Beyond TGF-β

Elisa Rossi 1,2*, Carmelo Bernabeu 3 and David M. Smadja 1,2,4,5

1 Université Paris Descartes, Sorbonne Paris Cité, Paris, France, 2 Inserm UMR-S1140, Paris, France, 3 Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain, 4 Department of Hematology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France, 5 Laboratory of Biosurgical Research, Carpentier Foundation, Hôpital Européen Georges Pompidou, Paris, France

Endoglin (ENG) is a transmembrane expressed on endothelial cells that functions as a co-receptor for several ligands of the transforming growth factor beta (TGF-β) family. ENG is also a recognized marker of and mutations in the endoglin gene are responsible for Hereditary Hemorrhagic Telangiectasia (HHT) type 1, a vascular disease characterized by defective angiogenesis, arteriovenous malformations,

Edited by: telangiectasia, and epistaxis. In addition to its involvement in the TGF-β family signaling Peter S. Steyger, pathways, several lines of evidence suggest that the extracellular domain of ENG has Oregon Health and Science University, a role in integrin-mediated adhesion via its RGD motif. Indeed, we have described United States a role for endothelial ENG in leukocyte trafficking and extravasation via its binding to Reviewed by: Eleni Papakonstantinou, leukocyte integrins. We have also found that ENG is involved in vasculogenic properties of Aristotle University of Thessaloniki, endothelial progenitor cells known as endothelial colony forming cells (ECFCs). Moreover, Greece Valerie Kouskoff, the binding of endothelial ENG to platelet integrins regulate the resistance to shear during University of Manchester, platelet- interactions under inflammatory conditions. Because of the need for United Kingdom more effective treatments in HHT and the involvement of ENG in angiogenesis, current Michelle Letarte, Hospital for Sick Children, Canada studies are aimed at identifying novel biological functions of ENG which could serve as *Correspondence: a therapeutic target. This review focuses on the interaction between ENG and integrins Elisa Rossi with the aim to better understand the role of this in blood vessel formation driven [email protected] by progenitor and mature endothelial cells.

Specialty section: Keywords: endoglin, endothelial progenitors, EPC, ECFCs, HHT1, integrins, TGF-β This article was submitted to Translational Medicine, a section of the journal Frontiers in Medicine ENDOGLIN STRUCTURE AND FUNCTION Received: 27 June 2018 Endoglin (ENG; also known as CD105) is a type I transmembrane glycoprotein predominantly Accepted: 14 January 2019 expressed in endothelial cells (ECs) (1, 2) and endothelial colony-forming cells (ECFCs) Published: 30 January 2019 (3). It is known as an essential co-receptor for the transforming growth factor β (TGF- Citation: β) family, playing an important role in angiogenesis. At the cell surface, ENG associates Rossi E, Bernabeu C and Smadja DM with TGF-β type I receptors, including the ECs-specific ALK1 (-like kinase (2019) Endoglin as an Adhesion β Molecule in Mature and Progenitor 1) and the ubiquitous ALK5, as well as to a TGF- type II receptor. In this receptor Endothelial Cells: A Function Beyond complex, ENG and ALK1 are able to bind to bone morphogenetic protein 9 (BMP9; TGF-β. Front. Med. 6:10. also known as growth differentiation factor 2 [GDF2]), a member of the TGF-β family, doi: 10.3389/fmed.2019.00010 with much higher affinity than to TGF-β1 and mediate its proliferation signal (4–7).

Frontiers in Medicine | www.frontiersin.org 1 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors

Upon ligand binding, the TGF-β signaling receptor complex VASCULAR PATHOPHYSIOLOGY OF phosphorylates members of the Smad family of transcription ENDOGLIN IN HEREDITARY HEMORRAGIC factors which, in turn, undergo nuclear translocation to regulate TELANGECTASIA (HHT) (Figure 1A). In addition to the membrane bound form of endoglin, Hereditary Hemorrhagic Telangiectasia (HHT) is an a circulating form of the ENG ectodomain, was found to autosomal dominant disease with a prevalence of one case be released upon a C-terminal cleavage of ENG by matrix per 5,000–8,000 individuals (25, 26). HHT patients develop metalloproteinase 14 (MMP-14) (8, 9). Increased levels of muco-cutaneous lesions known as telangiectasia in the nose, circulating endoglin have been detected at early stages of mouth, and gastrointestinal tract, as well as larger AVMs preeclampsia (5, 10) in hypertensive or diabetic patients (11) in major organs such as the lung, liver, and brain (25). The and in certain cancer patients (12), suggesting its role as telangiectasia are comprised of fragile vessels that are susceptible a predictive biomarker in these pathologies. Interestingly, to rupture and hemorrhage, which means that HHT patients can a detailed characterization of plasma from women with suffer recurrent anemia following frequent and severe bleeding preeclampsia has revealed that circulating endoglin can be episodes. Mutations in the endoglin gene (ENG) are responsible complexed within exosomes, rather than as individual soluble for HHT type 1 (HHT1) (27), while mutations in activing endoglin (13). receptor-like kinase (ACVRL1 alias ALK1) are responsible for A pioneer article on human ENG described that this HHT2 (2, 27). Taken together, HHT1 and HHT2 account for glycoprotein is present on ECs as a dimer (two subunits more than 80% of all HHT cases, whereas a small number of of ∼90-kDa), each displaying the tripeptide arginine-glycine- patients show mutations in SMAD4 (MADH4) or in BMP9 aspartic acid (RGD) in the extracellular region of the protein gene (GDF2), which are responsible for less common HHT (1). The RGD motif and homologous sequences in mouse variants such as a combined syndrome with juvenile polyposis and pig are recognition motifs for cell surface integrins (JP-HHT) and HHT5, respectively (28, 29). Remarkably, all present in such as fibronectin, of these genes code for proteins involved in the TGF-β family tenascin, thrombospondin, vitronectin, von Willebrand factor signaling pathways (30). Nowadays, the most widely accepted as well as fibrinogen and prothrombin (14–16). Integrin- hypotheses for AVMs formation in HHT1 and HHT2 involves mediated adhesion is involved in hemostasis, thrombosis, and a “first hit” (gene haploinsufficiency) based on the loss of inflammation, processes in which the endothelium plays a one functional allele Eng+/− (HHT1) or ALK1+/− (HHT2), critical role. Therefore, the presence of the RGD motif within which causes a 50% reduction in protein expression, followed a hydrophilic environment (1) and its accessibility in the 3D by a “second hit” based on an angiogenic trigger such as structure of ENG (7), clearly suggest the involvement of ENG inflammation, hypoxia or vascular injury (31). Together, these in integrin binding, as postulated in early studies (17, 18). In events may induce a deficient endothelial function of ENG fact, endothelial endoglin binds to leukocyte integrins, allowing that could result in HHT vascular lesions. The relevant role leukocyte extravasation (19) suggesting a possible novel function of endoglin in the pathophysiology of the vascular system for ENG independent of TGF-β signaling (Figures 1B,C). Several is illustrated by the phenotype of different HHT1 animal lines of evidence support the role of ENG in leukocyte trafficking: models (32). While endoglin heterozygous mice are viable (i) ENG expression is markedly up regulated in endothelial cells and reproduce the vascular phenotype of HHT patients, total of inflamed tissues with an associated inflammatory cell infiltrate loss of ENG expression leads in mice to cardiovascular defects (19); (ii) ENG is up-regulated in the post-ischemic kidney and and embryonic death by mid-gestation (33–35). A conditional ENG-haploinsufficient mice are protected from renal ischemia- knock out mouse model for HHT1 has revealed that the reperfusion injury, due to a reduction of cellular inflammatory arteriovenous malformations (AVMs), induced upon ENG responses (20); (iii) Arterial, venous, and capillary endothelia in loss, appear to be the result of delayed vascular remodeling lymphoid organs are highly reactive with anti-ENG and inappropriate ECs proliferation responses (36). In ENG and a marked staining pattern is observed in high endothelial null mice, vasculogenesis does not seem affected, suggesting venules (21); and (iv) Although ENG is present throughout that ENG is not required for initial differentiation of ECs or the vascular endothelium, its expression, as compared to veins formation of a primitive vascular plexus. The yolk sac of Eng−/− or arteries, is stronger in capillaries, where most leukocyte embryos has enlarged fragile vessels, while the embryo proper infiltration to organs occurs. shows cardiac cushion defects and delayed maturation of major In addition to leukocytes, other cell types present in the vessels (34). The reason why the total loss of ENG expression circulatory system have also revealed an integrin-mediated leads to cardiovascular defects and embryonic death is not adhesion activity toward endothelial endoglin (22–24) (Figures 1D,E). Thus, endoglin plays a role in integrin-mediated completely understood. Some authors have postulated that adhesion of vascular mural cells to endothelium (23). Also, a the primary defect is in the heart (37), while others suggest new role for endoglin in αIIbβ3 integrin-mediated adhesion of that the loss of smooth muscle cells (SMCs) coverage of the −/− platelets to the endothelium, conferring resistance of adherent endothelium could play a decisive role in the lethality of Eng platelets to detachment has been described (24)(Figure 1E). animals (34).

Frontiers in Medicine | www.frontiersin.org 2 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors

FIGURE 1 | Hypothetical model of Endoglin and its role as TGF-β co-receptor and adhesion molecule in different physiological contexts. (A) Canonical pathway that implicates Eng as a co-receptor of TGF-β in ECs. Bone morphogenetic protein 9 (BMP9), and other members of the TGF-β family, bind to an EC receptor complex composed by the type I (R-I) receptor named ALK1 and the type II (R-II) receptor, both exhibiting serine/threonine kinase activity, as well as the auxiliary receptor endoglin. Upon ligand binding, the R-II transphosphorylates ALK1, which subsequently propagates the signal by phosphorylating the receptor-regulated Smad (R-Smad) family of proteins Smad1/5/8. Phosphorylated R-Smads form heteromeric complexes with Smad4, translocating into the nucleus to regulate the transcriptional activity of different target genes. BMP9, Endoglin, ALK1 and Smad4 proteins are encoded by GDF2, ENG, ACVRL1 and MADH4 genes, whose pathogenic mutations give rise to HHT5, HHT1, HHT2, and JPHT, respectively (30). (B) Endothelial endoglin as an adhesion molecule involving its RGD sequence when binding to cell surface integrins from leukocytes, VMCs or platelets. (C) Leukocyte transmigration through the vessel endothelium. Under inflammatory conditions, different soluble factors are released leading to leukocyte adhesion and transmigration through ECs. This process is mediated, at least, by the interaction between leukocyte integrin α5β1 and endothelial endoglin involving the specific recognition of the RGD motif in Eng (22). (D) Adhesion between endothelial cells and vascular mural cells (VMCs). This intercellular association involves, at least, the interaction between integrin α5β1 in VMCs and endothelial endoglin via the specific recognition of the RGD motif in Eng (23). (E) Platelet-dependent hemostasis. Endothelial endoglin via its RGD binds to platelet integrins αIIbβ3 and α5β1, contributing to stabilize platelets adhesion to endothelium (24).

ENDOGLIN INTERACTING PARTNERS IN Ras-related C3 botulinum toxin substrate 2 (Rac-2), involved THE VASCULATURE in endothelial barrier maintenance, cytoskeletal remodeling and cell migration (44). Using in vitro ECs lines derived Previous studies reported that endothelial ENG is associated with from ENG homozygous null, heterozygous, and control mouse several proteins critically involved in endothelial cell adhesion, embryos, Jerkic and Letarte have reported that ENG-deficient proliferation, migration, angiogenesis, and vascular permeability, mouse embryonic ECs are hyper-permeable and unresponsive including integrins (38), zyxin (39), zyxin-related protein 1 to stimulation by VEGF and TGF-β1, factors that regulate (ZRP-1) (40), VEGF receptor type 2 (VEGFR-2) (41), beta- vascular permeability (45). Interestingly, VE-cadherin is barely arrestin 2 (42), and vascular endothelial-cadherin (VE-cadherin) detected in ENG-deficient ECs, a finding that could explain (43). It has been shown that VE-cadherin interacts with several the EC junction destabilization and impairment of TGF- components of the TGF-β receptor complex, including ENG, β signaling. Furthermore, permeability alterations in Eng+/− ALK1, and the TGF-β type II receptor TGFBR2 in adherents mice are in line with findings in patients with Hereditary junctions. Of note, ENG, TGFBR2, and ALK1 also interact Hemorrhagic Telangiectasia type 1 (HHT1). Thus, ECs from with p21-activated kinase (PAK-1), GTP-binding proteins, and HHT1 patients show a disorganized actin cytoskeleton prone to

Frontiers in Medicine | www.frontiersin.org 3 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors cell breaking with changes in shear stress and blood pressure and in vivo and they are involved in blood vessel formation and (46). This reorganization of actin filaments in HHT1 might vascular repair (58, 59). For example, upon vascular injury, tissue lead to vessel hemorrhages and eventual disappearance of the perfusion of blood flow involves not only angiogenic sprouting capillary network, as reported in this disorder. In line with the of ECs from nearby intact vessels, but also the recruitment pathogenic hypothesis of an endothelial dysfunction in HHT1, circulating ECFCs allowing the formation of new blood vessels the involvement of ENG in integrin-mediated trans-endothelial (60). Acting at the interface between blood and tissues, ECFCs leukocyte trafficking was proposed (31). Accordingly, following have a remarkable ability for migration and proliferation, inflammation triggering, the extracellular region of ENG binds to being key cells in angiogenesis and vascular remodeling. It leukocyte integrin α5β1 and promotes leukocyte transmigration is therefore not surprising that perturbations in these critical (31). In contrast, a different experimental approach showed an ECFCs functions contribute to several vascular pathologies. increased gut inflammation and myeloid infiltration in colitis Accordingly, ECFCs have opened a new area for treatment Eng+/− vs. control mice (47). Overall, this novel role of ENG may of cardiovascular diseases using cell therapy, advancing in the contribute to a better understanding of the inflammation process knowledge of vessel reconstruction ability in postnatal life. In and the high incidence of infections in HHT patients (48). this line, much effort has been recently devoted to identify Because pericytes embrace the endothelium to regulate the blood novel molecular targets within ECFCs able to enhance their retinal barrier, the role of endothelial endoglin in this context angiogenic potential. ECFCs are positive for endoglin (CD105), has also been analyzed. Thus, vascular permeability studies VE-cadherin (CD144), CD31, VEGFR2 (KDR), EGFL7, and carried out in vivo showed that retinas of Eng+/− heterozygous CD146, and negative for CD45 and CD14 (58, 59). Among these, mice displayed higher retinal permeability than that of Eng+/+ ENG is emerging as an interesting therapeutic target based on its mice, suggesting a destabilization of the endothelial barrier involvement in angiogenesis and vascular remodeling (46, 61). function due to ENG haploinsufficiency (23). The importance Many strategies of therapeutic revascularization, based on of the interaction between ECs and vascular mural cells is the administration of growth factors or stem/progenitor cells also underlined by studies showing that thalidomide stabilizes from diverse sources including EPCs, have been proposed the vasculature in a mouse model of HHT1 (49). Indeed, and are currently being tested in patients with peripheral pericyte recruitment and stabilization appears as a therapeutic arterial disease or cardiac diseases (62). Some pretreatment of strategy to reduce the severity of epistaxis in HHT (50). Thus, ECFCs with erythropoietin (63), fucoidan (64), soluble CD146 ENG, directly, or as a component of the TGF-β receptor (65), tumor necrosis factor-α (TNF-α) (66), or platelet lysates complex may regulate ECs integrity, whereas its absence may (67, 68) have also been proposed to improve the therapeutic result in vascular hyper-permeability, thus contributing to the efficacy of in vivo administered ECFCs. Interestingly, integrins fetal lethality associated with the homozygous null genotype represent a major molecular determinant of EPCs function. More (39, 40). specifically, integrin α4β1 is a key regulator of EPCs retention Although this review focuses on the role of endoglin in and/or mobilization from the bone marrow, while integrins ECs, it is worth mentioning that endoglin is also expressed α5β1, α6β1, αvβ3, and αvβ5 are involved in EPCs homing, at lower levels in other cell types, some of which present invasion, differentiation and paracrine factor production (69). in the vasculature as vascular SMCs, activated monocytes or The involvement of ECFCs in angiogenesis has been shown to mesenchymal cells (18, 51–53). However, a review on the be essential for ischemic disease recovery (70, 71). Furthermore, endoglin role in these cell types deserves an independent the differentiation of mesenchymal stem cells (MSCs) into mural study. cells appears to be stimulated by their co-culture with ECFCs, whereas overexpression of the Notch ligand Jagged1 in ECs further enhanced the differentiation of MSC into pericytes ENDOGLIN AND ENDOTHELIAL (72). This cooperation prompted some authors to postulate COLONIES FORMING CELLS (ECFCS) an improvement of ECFCs efficiency by co-injecting ECFCs and SMC progenitors (73). Therefore, a combined treatment of The formation of blood vessels from mesoderm is driven by the ECFCs with mesenchymal stem/progenitor cells (MSCs/MPCs) recruitment of undifferentiated cells which can differentiate into appears to operate synergistically, enhancing neovascularization endothelial lineage. In the adult, populations of bone marrow- compared to either individual cell population (74, 75). Recently, derived endothelial progenitor cells (EPCs) are mobilized into we proposed a role for ENG in the ECFCs-MSCs interplay the circulation by stimuli such as estrogen and VEGF (54). involved in the revascularization process (61). Indeed, ENG Interestingly, human EPCs could derive from very small silencing in ECFCs markedly inhibited ECFCs adhesion to MSCs embryonic like stem cells (55) that can also give rise to in vitro, without affecting MSCs differentiation into perivascular hematopoietic cells (56). Two populations of EPCs have cells or other mesenchymal lineages. Mesenchymal stem cells been differentially characterized from circulating endothelial (MSCs) increase muscle recovery of ECFC in HLI model and progenitors: (i) early EPCs that exhibit a hematopoietic profile we found that ENG-silenced ECFCs co-injected with MSCs in with a genomic fingerprint similar to monocytes; and (ii) ECFCs, mice abolish beneficial effects of MSCs leading to decreased vessel also known as blood outgrowth endothelial cells (BOECs), which density and foot perfusion upon ischemia. Our results suggest are considered late EPCs with an endothelial-like genomic profile ENG involvement in the crosstalk between ECFCs and MSCs, (57). ECFCs display an intrinsic tube forming capacity in vitro leading to vessel formation and stabilization (51)(Figures 2A,B).

Frontiers in Medicine | www.frontiersin.org 4 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors

FIGURE 2 | Endoglin in ECFCs. (A) Role of endoglin matrigel vascularization in vivo. Matrigel plugs were mixed with either ECFCs treated with control siRNA plus mesenchymal stem cells (MSCs) (left) or with ECFCs treated with endoglin specific siRNA plus MSCs (right). The Matrigel mixture was injected into nude mice and the number of vessels was analyzed after 1 week. Control plugs display a marked vascularization with functional vessels (presence of erythrocytes). Plugs with endoglin-silenced ECFCs display less vascularization than controls, suggesting an important role for this protein in cell adhesion (23). (B) MSCs combined with ECFCs, accelerate muscle recovery in a mouse model of hind limb ischemia, through an endoglin-dependent mechanism. After femoral ligature and retro-orbital injection of ECFC+MSC Doppler analysis shows a revascularization induced by co-injection of ECFCs plus MSCs (left). This synergistic effect is abolished when endoglin is previously silenced in ECFCs (right) (61). (C) Mechanisms involved in the synergy between ECFCs and MSCs. (i) Mutual paracrine effect between ECFCs and MSCs involving growth factors like VEGF and PDGFBB (76, 77). (ii) ECFCs-induced differentiation of MSCs into perivascular cells via Notch-Jagged1 (72) (iii) Adhesion between ECFCs and MSCs involving endoglin (61).

As postulated in Figure 2C, three steps may be involved Thus, the ENG cytoplasmic domain binds to zyxin and ZRP1, during the formation of stable vessels by ECFC and MSC which concentrate at focal adhesions within actin polymerization and/or perivascular cells: (i) Mutual paracrine effects by VEGF points. Consequently, the decrease of ENG levels in HHT1 and/or PDGF-BB (76, 77). (ii) ECFCs-driven perivascular ECFCs could be related to the cytoskeleton alteration. maturation of MSC and/or SMC progenitors via the notch- dependent pathway; and (iii) Adhesion between ECFCs CONCLUSION and perivascular cells in an ENG-dependent manner. The relevance of endothelial ENG adhesive properties in Blood vessel formation and remodeling are essential processes mature vessel formation in mice (61) is in agreement with in the maintenance of tissue homeostasis and function, a previous report on the active role of human ENG in the and therefore, their alteration causes a variety of pathologic adhesion between mature ECs and vascular mural cells conditions. ENG involvement in the function of mature and (23). progenitor endothelial cells is a hot topic not completely Relevance of ENG in ECFC has been demonstrated also understood and developed yet. Recent findings underline in ECFCs isolated directly from HHT1 patients. Indeed, these the importance of ENG not only as a co-receptor for several ECFCs have disorganized and depolymerized actin fibers and ligands of the TGF-β family, but also as a molecule involved impaired tube formation, as compared to healthy ECFCs (46). in cell adhesion, which can regulate ECFCs behavior in The molecular basis for the abnormal behavior of HHT1 ECFCs the context of angiogenic processes. It is recognized that appears to be mediated, at least in part, by the regulatory in mature ECs, ENG plays a key role in angiogenesis and role of endoglin and we can speculate via its interaction blood vessel homeostasis, becoming a potential therapeutic with zyxin family members involved in the actin cytoskeletal target for pro- and anti-angiogenic approaches in the organization as it was proposed for endothelial cells (39, 40). treatment of diseases such as HHT, cancer, preeclampsia,

Frontiers in Medicine | www.frontiersin.org 5 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors diabetes complications or post-ischemic disease. While ENG and platelets, independently or in parallel to its TGF-β co- expression is relatively low in quiescent ECs, it is upregulated receptor role. in the active endothelium involved in angiogenesis and While evidence-based therapeutics are coming into play in vessel remodeling and permeability. In these processes, the traditionally empirical base for endoglin related protein endothelial ENG regulates ECs proliferation, migration, and derivatives, future studies of ENG in endothelial progenitor cells actin cytoskeletal organization, leukocyte extravasation, and may pave the way for a better understanding of its function in the interaction with mural cells. In addition, a role for ENG in vascular system and for the development and understanding of platelet-endothelium interaction, including the resistance of the use of these immature cells as a cell therapy product. adherent platelets to shear under inflammatory conditions, was also recently proposed, opening new perspectives on ENG AUTHOR CONTRIBUTIONS functions. This review highlights the emerging roles of endothelial ER wrote the manuscript. CB provided helpful suggestions and ENG as a cell adhesion molecule in mature and progenitor contributed to writing the manuscript. DS wrote the endothelial cells interacting with vascular mural cells, leukocytes, manuscript and provided funding support.

REFERENCES 15. Luo B-H, Carman CV, Springer TA. Structural basis of integrin regulation and signaling. Annu Rev Immunol. (2007) 25:619–47. 1. Gougos A, Letarte M. Primary structure of endoglin, an RGD-containing doi: 10.1146/annurev.immunol.25.022106.141618 glycoprotein of human endothelial cells. J Biol Chem. (1990) 265:8361–4. 16. Eto K, Puzon-McLaughlin W, Sheppard D, Sehara-Fujisawa A, Zhang X- 2. López-Novoa JM, Bernabeu C. The physiological role of endoglin in the P, Takada Y. RGD-independent binding of integrin α9β1 to the ADAM- cardiovascular system. Am J Physiol Heart Circ Physiol. (2010) 299:H959–74. 12 and−15 disintegrin domains mediates cell-cell interaction. J Biol Chem. doi: 10.1152/ajpheart.01251.2009 (2000) 275:34922–30. doi: 10.1074/jbc.M001953200 3. Campioni D, Zauli G, Gambetti S, Campo G, Cuneo A, Ferrari R, et al. 17. Gougos A, St Jacques S, Greaves A, O’Connell PJ, d’Apice AJ, Bühring HJ, In vitro characterization of circulating endothelial progenitor cells isolated et al. Identification of distinct epitopes of endoglin, an RGD-containing from patients with acute coronary syndrome. PLoS ONE (2013) 8:e56377. glycoprotein of endothelial cells, leukemic cells, and syncytiotrophoblasts. Int doi: 10.1371/journal.pone.0056377 Immunol. (1992) 4:83–92. 4. Alt A, Miguel-Romero L, Donderis J, Aristorena M, Blanco FJ, Round A, et 18. Lastres P,Bellon T, Cabañas C, Sanchez-Madrid F, Acevedo A, Gougos A, et al. al. Structural and functional insights into endoglin ligand recognition and Regulated expression on human of endoglin, an Arg-Gly-Asp- binding. PLoS ONE (2012) 7:e29948. doi: 10.1371/journal.pone.0029948 containing surface antigen. Eur J Immunol. (1992) 22:393–7. 5. Gregory AL, Xu G, Sotov V, Letarte M. Review: the enigmatic role of endoglin 19. Torsney E, Charlton R, Parums D, Collis M, Arthur HM. Inducible expression in the placenta. Placenta (2014) 35:S93–9. doi: 10.1016/j.placenta.2013.10.020 of human endoglin during inflammation and wound healing in vivo. Inflamm 6. Li W, Salmon RM, Jiang H, Morrell NW. Regulation of the ALK1 Res Off J Eur Histamine Res Soc Al. (2002) 51:464–70. doi: 10.1007/PL000 ligands, BMP9 and BMP10. Biochem Soc Trans. (2016) 44:1135–41. 12413 doi: 10.1042/BST20160083 20. Docherty NG, López-Novoa JM, Arevalo M, Düwel A, Rodriguez-Peña A, 7. Saito T, Bokhove M, Croci R, Zamora-Caballero S, Han L, Letarte Pérez-Barriocanal F, et al. Endoglin regulates renal ischaemia-reperfusion M, et al. Structural basis of the human endoglin-BMP9 interaction: injury. Nephrol Dial Transpl. (2006) 21:2106–19. doi: 10.1093/ndt/gfl179 insights into BMP signaling and HHT1. Cell Rep. (2017) 19:1917–28. 21. Dagdeviren A, Müftüoglu SF,Cakar AN, Ors U. Endoglin (CD 105) expression doi: 10.1016/j.celrep.2017.05.011 in human lymphoid organs and placenta. Ann Anat Anat Anz. (1998) 8. Hawinkels LJAC, Kuiper P, Wiercinska E, Verspaget HW, Liu Z, Pardali 180:461–9. E, et al. Matrix metalloproteinase-14 (MT1-MMP)-mediated endoglin 22. Rossi E, Sanz-Rodriguez F, Eleno N, Düwell A, Blanco FJ, Langa shedding inhibits tumor angiogenesis. Cancer Res. (2010) 70:4141–50. C, et al. Endothelial endoglin is involved in inflammation: role in doi: 10.1158/0008-5472.CAN-09-4466 leukocyte adhesion and transmigration. Blood (2013) 121:403–15. 9. Valbuena-Diez AC, Blanco FJ, Oujo B, Langa C, Gonzalez-Nuñez M, Llano doi: 10.1182/blood-2012-06-435347 E, et al. Oxysterol-induced soluble endoglin release and its involvement 23. Rossi E, Smadja DM, Boscolo E, Langa C, Arevalo MA, Pericacho M, et al. in hypertension clinical perspective. Circulation (2012) 126:2612–24. Endoglin regulates mural cell adhesion in the circulatory system. Cell Mol Life doi: 10.1161/CIRCULATIONAHA.112.101261 Sci. (2016) 73:1715–39. doi: 10.1007/s00018-015-2099-4 10. Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, et al. 24. Rossi E, Pericacho M, Bachelot-Loza C, Pidard D, Gaussem P, Poirault- Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. Chassac S, et al. Human endoglin as a potential new partner involved (2006) 12:642–9. doi: 10.1038/nm1429 in platelet–endothelium interactions. Cell Mol Life Sci. (2018) 75:1269–84. 11. Blázquez-Medela AM, García-Ortiz L, Gómez-Marcos MA, Recio-Rodríguez doi: 10.1007/s00018-017-2694-7 JI, Sánchez-Rodríguez A, López-Novoa JM, et al. Increased plasma 25. Shovlin CL. Hereditary haemorrhagic telangiectasia: pathophysiology, soluble endoglin levels as an indicator of cardiovascular alterations in diagnosis and treatment. Blood Rev. (2010) 24:203–19. hypertensive and diabetic patients. BMC Med. (2010) 8:86. doi: 10.1186/1741- doi: 10.1016/j.blre.2010.07.001 7015-8-86 26. Faughnan ME, Palda VA, Garcia-Tsao G, Geisthoff UW, McDonald J, 12. Bernabeu C, Lopez-Novoa JM, Quintanilla M. The emerging role of TGF- Proctor DD, et al. International guidelines for the diagnosis and management β superfamily coreceptors in cancer. Biochim Biophys Acta BBA (2009) of hereditary haemorrhagic telangiectasia. J Med Genet. (2011) 48:73–87. 1792:954–73. doi: 10.1016/j.bbadis.2009.07.003 doi: 10.1136/jmg.2009.069013 13. Ermini L, Ausman J, Melland-Smith M, Yeganeh B, Rolfo A, Litvack ML, et 27. McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Baldwin MA, Jackson al. A single sphingomyelin species promotes exosomal release of endoglin CE, et al. Endoglin, a TGF-β binding protein of endothelial cells, is the gene into the maternal circulation in preeclampsia. Sci Rep. (2017) 7:12172. for hereditary haemorrhagic telangiectasia type 1. Nat Genet. (1994) 8:345–51. doi: 10.1038/s41598-017-12491-4 doi: 10.1038/ng1294-345 14. Ruoslahti E. RGD and other recognition sequences for integrins. Annu Rev 28. Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Tejpar S, et al. Cell Dev Biol. (1996) 12:697–715. doi: 10.1146/annurev.cellbio.12.1.697 A combined syndrome of juvenile polyposis and hereditary haemorrhagic

Frontiers in Medicine | www.frontiersin.org 6 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors

telangiectasia associated with mutations in MADH4 (SMAD4). Lancet (2004) 47. Peter MR, Jerkic M, Sotov V, Douda DN, Ardelean DS, Ghamami N, 363:852–9. doi: 10.1016/S0140-6736(04)15732-2 et al. Impaired resolution of inflammation in the endoglin heterozygous 29. Wooderchak-Donahue WL, McDonald J, O’Fallon B, Upton PD, Li W, mouse model of chronic colitis. Mediators Inflamm. (2014) 2014:767185. Roman BL, et al. BMP9 mutations cause a vascular-anomaly syndrome with doi: 10.1155/2014/767185 phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum 48. Dupuis-Girod S, Giraud S, Decullier E, Lesca G, Cottin V, Faure F, et al. Genet. (2013) 93:530–7. Hemorrhagic hereditary telangiectasia (Rendu-Osler disease) and infectious 30. Ruiz-Llorente L, Gallardo-Vara E, Rossi E, Smadja DM, Botella LM, diseases: an underestimated association. Clin Infect Dis Off Publ Infect Dis Soc Bernabeu C. Endoglin and alk1 as therapeutic targets for hereditary Am. (2007) 44:841–5. doi: 10.1086/511645 hemorrhagic telangiectasia. Expert Opin Ther Targets (2017) 21:933–47. 49. Lebrin F, Srun S, Raymond K, Martin S, Brink S van den, Freitas C, doi: 10.1080/14728222.2017.1365839 et al. Thalidomide stimulates vessel maturation and reduces epistaxis in 31. Rossi E, Lopez-Novoa JM, Bernabeu C. Endoglin involvement in integrin- individuals with hereditary hemorrhagic telangiectasia. Nat Med. (2010) mediated cell adhesion as a putative pathogenic mechanism in hereditary 16:420–8. doi: 10.1038/nm.2131 hemorrhagic telangiectasia type 1 (HHT1). Front Genet. (2015) 5:457. 50. Thalgott J, Dos-Santos-Luis D, Lebrin F. Pericytes as targets in doi: 10.3389/fgene.2014.00457 hereditary hemorrhagic telangiectasia. Front Genet. (2015) 6:37. 32. Tual-Chalot S, Oh SP, Arthur HM. Mouse models of hereditary hemorrhagic doi: 10.3389/fgene.2015.00037 telangiectasia: recent advances and future challenges. Front Genet. (2015) 6:25. 51. Guerrero-Esteo M, Lastres P, Letamendía A, Pérez-Alvarez MJ, Langa C, doi: 10.3389/fgene.2015.00025 López LA, et al. Endoglin overexpression modulates cellular morphology, 33. Bourdeau A, Dumont DJ, Letarte M. A murine model of hereditary migration, and adhesion of mouse fibroblasts. Eur J Cell Biol. (1999) 78: hemorrhagic telangiectasia. J Clin Invest. (1999) 104:1343–51. 614–23. doi: 10.1172/JCI8088 52. O’Connor JC, Farach-Carson MC, Schneider CJ, Carson DD. Coculture with 34. Li DY, Sorensen LK, Brooke BS, Urness LD, Davis EC, Taylor DG, et al. prostate cancer cells alters endoglin expression and attenuates transforming Defective Angiogenesis in mice lacking endoglin. Science (1999) 284:1534–7. growth factor-beta signaling in reactive bone marrow stromal cells. Mol doi: 10.1126/science.284.5419.1534 Cancer Res. (2007) 5:585–603. doi: 10.1158/1541-7786.MCR-06-0408 35. Arthur HM, Ure J, Smith AJH, Renforth G, Wilson DI, Torsney E, et 53. Tian H, Ketova T, Hardy D, Xu X, Gao X, Zijlstra A, et al. Endoglin al. Endoglin, an Ancillary TGFβ receptor, is required for extraembryonic mediates vascular maturation by promoting vascular smooth muscle cell angiogenesis and plays a key role in heart development. Dev Biol. (2000) migration and spreading. Arterioscler Thromb Vasc Biol. (2017) 37:1115–26. 217:42–53. doi: 10.1006/dbio.1999.9534 doi: 10.1161/ATVBAHA.116.308859 36. Mahmoud M, Allinson KR, Zhai Z, Oakenfull R, Ghandi P, Adams RH, et al. 54. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, et al. Impaired Pathogenesis of arteriovenous malformations in the absence of endoglin. Circ recruitment of bone-marrow–derived endothelial and hematopoietic Res. (2010) 106:1425–33. doi: 10.1161/CIRCRESAHA.109.211037 progenitor cells blocks tumor angiogenesis and growth. Nat Med. (2001) 37. Nomura-Kitabayashi A, Anderson GA, Sleep G, Mena J, Karabegovic A, 7:1194–201. doi: 10.1038/nm1101-1194 Karamath S, et al. Endoglin is dispensable for angiogenesis, but required for 55. Guerin CL, Loyer X, Vilar J, Cras A, Mirault T, Gaussem P, et al. Bone- endocardial cushion formation in the midgestation mouse embryo. Dev Biol. marrow-derived very small embryonic-like stem cells in patients with critical (2009) 335:66–77. doi: 10.1016/j.ydbio.2009.08.016 leg ischaemia: evidence of vasculogenic potential. Thromb Haemost. (2015) 38. Tian H, Mythreye K, Golzio C, Katsanis N, Blobe GC. Endoglin mediates 113:1084–94. doi: 10.1160/TH14-09-0748 fibronectin/α5β1 integrin and TGF-β pathway crosstalk in endothelial cells. 56. Smadja DM. Bone marrow very small embryonic-like stem cells: new EMBO J. (2012) 31:3885–900. doi: 10.1038/emboj.2012.246 generation of autologous cell therapy soon ready for prime time? 39. Conley BA, Koleva R, Smith JD, Kacer D, Zhang D, Bernabéu C, et Rev. (2017) 13:198–201. doi: 10.1007/s12015-017-9718-4 al. Endoglin controls cell migration and composition of focal adhesions 57. Smadja DM, Bièche I, Silvestre J-S, Germain S, Cornet A, Laurendeau function of the cytosolic domain. J Biol Chem. (2004) 279:27440–9. I, et al. Bone morphogenetic proteins 2 and 4 are selectively doi: 10.1074/jbc.M312561200 expressed by late outgrowth endothelial progenitor cells and promote 40. Sanz-Rodriguez F, Guerrero-Esteo M, Botella L-M, Banville D, Vary CPH, neoangiogenesis. Arterioscler Thromb Vasc Biol. (2008) 28:2137–43. Bernabéu C. Endoglin regulates cytoskeletal organization through binding doi: 10.1161/ATVBAHA.108.168815 to ZRP-1, a member of the Lim family of proteins. J Biol Chem. (2004) 58. d’Audigier C, Susen S, Blandinieres A, Mattot V, Saubamea B, Rossi E, et al. 279:32858–68. doi: 10.1074/jbc.M400843200 Egfl7 represses the vasculogenic potential of human endothelial progenitor 41. Tian H, Huang JJ, Golzio C, Gao X, Hector-Greene M, Katsanis N, et al. cells. Stem Cell Rev. (2018) 14:82–91. doi: 10.1007/s12015-017-9775-8 Endoglin interacts with VEGFR2 to promote angiogenesis. FASEB J Off Publ 59. Medina RJ, Barber CL, Sabatier F, Dignat-George F, Melero-Martin Fed Am Soc Exp Biol. (2018) 32:2934–49. doi: 10.1096/fj.201700867RR JM, Khosrotehrani K, et al. Endothelial progenitors: a consensus 42. Lee NY, Blobe GC. The Interaction of endoglin with β-Arrestin2 statement on nomenclature. Stem Cells Transl Med. (2017) 6:1316–20. regulates transforming growth factor-β-mediated ERK activation and doi: 10.1002/sctm.16-0360 migration in endothelial cells. J Biol Chem. (2007) 282:21507–17. 60. Banno K, Yoder MC. Tissue regeneration using endothelial colony-forming doi: 10.1074/jbc.M700176200 cells: promising cells for vascular repair. Pediatr Res. (2018) 83:283–90. 43. Rudini N, Felici A, Giampietro C, Lampugnani M, Corada M, Swirsding K, doi: 10.1038/pr.2017.231 et al. VE-cadherin is a critical endothelial regulator of TGF-beta signalling. 61. Rossi E, Goyard C, Cras A, Dizier B, Bacha N, Lokajczyk A, et al. Co-injection EMBO J. (2008) 27:993–1004. doi: 10.1038/emboj.2008.46 of mesenchymal stem cells with endothelial progenitor cells accelerates muscle 44. Xu G, Barrios-Rodiles M, Jerkic M, Turinsky AL, Nadon R, Vera S, et al. Novel recovery in hind limb ischemia through an endoglin-dependent mechanism. protein interactions with endoglin and activin receptor-like kinase 1: potential Thromb Haemost. (2017) 117:1908–18. doi: 10.1160/TH17-01-0007 role in vascular networks. Mol Cell Proteomics MCP (2014) 13:489–502. 62. Silvestre J-S, Smadja DM, Lévy BI. Postischemic revascularization: from doi: 10.1074/mcp.M113.033464 cellular and molecular mechanisms to clinical applications. Physiol Rev. (2013) 45. Seghers L, de Vries MR, Pardali E, Hoefer IE, Hierck BP, ten Dijke 93:1743–802. doi: 10.1152/physrev.00006.2013 P ten, et al. Shear induced collateral artery growth modulated by 63. Bennis Y, Sarlon-Bartoli G, Guillet B, Lucas L, Pellegrini L, Velly L, et al. endoglin but not by ALK1. J Cell Mol Med. (2012) 16:2440–50. Priming of late endothelial progenitor cells with erythropoietin before doi: 10.1111/j.1582-4934.2012.01561.x transplantation requires the CD131 receptor subunit and enhances 46. Fernandez-L A, Sanzrodriguez F, Zarrabeitia R, Perezmolino A, their angiogenic potential. J Thromb Haemost. (2012) 10:1914–28. Hebbel R, Nguyen J, et al. Blood outgrowth endothelial cells from doi: 10.1111/j.1538-7836.2012.04835.x Hereditary Haemorrhagic Telangiectasia patients reveal abnormalities 64. Lee JH, Lee SH, Choi SH, Asahara T, Kwon S-M. The sulfated polysaccharide compatible with vascular lesions. Cardiovasc Res. (2005) 68:235–48. fucoidan rescues senescence of endothelial colony-forming cells for ischemic doi: 10.1016/j.cardiores.2005.06.009 repair. Stem Cells Dayt Ohio (2015) 33:1939–51. doi: 10.1002/stem.1973

Frontiers in Medicine | www.frontiersin.org 7 January 2019 | Volume 6 | Article 10 Rossi et al. Endoglin and Endothelial Progenitors

65. Stalin J, Harhouri K, Hubert L, Garrigue P, Nollet M, Essaadi A, et al. 73. Foubert P, Matrone G, Souttou B, Leré-Déan C, Barateau V, Plouët J, et al. Soluble CD146 boosts therapeutic effect of endothelial progenitors through Coadministration of endothelial and smooth muscle progenitor cells enhances proteolytic processing of short CD146 isoform. Cardiovasc Res. (2016) the efficiency of proangiogenic cell-based therapy. Circ Res. (2008) 103:751– 111:240–51. doi: 10.1093/cvr/cvw096 60. doi: 10.1161/CIRCRESAHA.108.175083. 66. Green LA, Njoku V, Mund J, Case J, Yoder M, Murphy MP, et 74. Lin R-Z, Moreno-Luna R, Zhou B, Pu WT, Melero-Martin JM. al. Endogenous transmembrane TNF-alpha protects against premature Equal modulation of endothelial cell function by four distinct tissue- senescence in endothelial colony forming cells. Circ Res. (2016) 118:1512–24. specific mesenchymal stem cells. Angiogenesis (2012) 15:443–55. doi: 10.1161/CIRCRESAHA.116.308332 doi: 10.1007/s10456-012-9272-2 67. Kim H, Prasain N, Vemula S, Ferkowicz MJ, Yoshimoto M, Voytik- 75. Kang K-T, Lin R-Z, Kuppermann D, Melero-Martin JM, Bischoff J. Harbin SL, et al. Human platelet lysate improves human cord blood Endothelial colony forming cells and mesenchymal progenitor cells form derived ECFC survival and vasculogenesis in three dimensional (3D) blood vessels and increase blood flow in ischemic muscle. Sci Rep. (2017) collagen matrices. Microvasc Res. (2015) 101:72–81. doi: 10.1016/j.mvr.2015. 7:770. doi: 10.1038/s41598-017-00809-1 06.006 76. Smadja DM, Levy M, Huang L, Rossi E, Blandinières A, Israel-Biet D, et al. 68. Tasev D, van Wijhe MH, Weijers EM, van Hinsbergh VWM, Koolwijk Treprostinil indirectly regulates endothelial colony forming cell angiogenic P. Long-term expansion in platelet lysate increases growth of peripheral properties by increasing VEGF-A produced by mesenchymal stem cells. blood-derived endothelial-colony forming cells and their growth Thromb Haemost. (2015) 114:735–47. doi: 10.1160/TH14-11-0907. factor-induced sprouting capacity. PLoS ONE (2015) 10:e0129935. 77. Lin R-Z, Moreno-Luna R, Li D, Jaminet S-C, Greene AK, Melero-Martin doi: 10.1371/journal.pone.0129935 JM. Human endothelial colony-forming cells serve as trophic mediators for 69. Caiado F, Dias S. Endothelial progenitor cells and integrins: adhesive needs. engraftment via paracrine signaling. Proc Natl Acad Fibrogenesis Tissue Repair (2012) 5:4. doi: 10.1186/1755-1536-5-4 Sci USA. (2014) 111:10137–42. doi: 10.1073/pnas.1405388111 70. Guerin CL, Rossi E, Saubamea B, Cras A, Mignon V, Silvestre J, et al. Human very small embryonic-like cells support vascular maturation and therapeutic Conflict of Interest Statement: The authors declare that the research was revascularization induced by endothelial progenitor cells. Stem Cell Rev Rep. conducted in the absence of any commercial or financial relationships that could (2017) 13:552–60. doi: 10.1007/s12015-017-9731-7 be construed as a potential conflict of interest. 71. Smadja DM. Bone marrow cell therapy in cardiovascular disease drives us slowly to a better identification of the active cell component. Stem Cell Res Copyright © 2019 Rossi, Bernabeu and Smadja. This is an open-access article Ther. (2014) 5:16. doi: 10.1186/scrt405 distributed under the terms of the Creative Commons Attribution License (CC BY). 72. Shafiee A, Patel J, Wong HY, Donovan P, Hutmacher DW, Fisk NM, et The use, distribution or reproduction in other forums is permitted, provided the al. Priming of endothelial colony-forming cells in a mesenchymal niche original author(s) and the copyright owner(s) are credited and that the original improves engraftment and vasculogenic potential by initiating mesenchymal publication in this journal is cited, in accordance with accepted academic practice. transition orchestrated by NOTCH signaling. FASEB J. (2017) 31:610–24. No use, distribution or reproduction is permitted which does not comply with these doi: 10.1096/fj.201600937 terms.

Frontiers in Medicine | www.frontiersin.org 8 January 2019 | Volume 6 | Article 10